Optimization of 1,2,4-Triazolopyridines as Inhibitors of Human 11β

May 22, 2014 - †Department of Medicinal Chemistry, ‡Department of Biology, §Lead Evaluation, ∥CADD, ⊥Department of Pharmaceutical Candidate O...
0 downloads 11 Views 930KB Size
Letter pubs.acs.org/acsmedchemlett

Optimization of 1,2,4-Triazolopyridines as Inhibitors of Human 11βHydroxysteroid Dehydrogenase Type 1 (11β-HSD-1) Jun Li,*,† Lawrence J. Kennedy,† Haixia Wang,† James J. Li,† Steven J. Walker,† Zhenqiu Hong,† Stephen P. O’Connor,† Akbar Nayeem,∥ Daniel M. Camac,# Paul E. Morin,# Steven Sheriff,# Mengmeng Wang,⊥ Timothy Harper,⊥ Rajasree Golla,§ Ramakrishna Seethala,§ Thomas Harrity,‡ Randolph P. Ponticiello,‡ Nathan N. Morgan,‡ Joseph R. Taylor,‡ Rachel Zebo,‡ David A. Gordon,‡ and Jeffrey A. Robl*,† †

Department of Medicinal Chemistry, ‡Department of Biology, §Lead Evaluation, ∥CADD, ⊥Department of Pharmaceutical Candidate Optimization, Bristol-Myers Squibb, Bldg 13, Princeton, New Jersey 08543-5400, United States # Protein Science & Structure, Research & Development, Bristol-Myers Squibb, Princeton, New Jersey 08543, United States S Supporting Information *

ABSTRACT: Small alkyl groups and spirocyclic-aromatic rings directly attached to the left side and right side of the 1,2,4-triazolopyridines (TZP), respectively, were found to be potent and selective inhibitors of human 11βhydroxysteroid dehydrogenase-type 1 (11β-HSD-1) enzyme. 3-(1-(4Chlorophenyl)cyclopropyl)-8-cyclopropyl-[1,2,4]triazolo[4,3-a]pyridine (9f) was identified as a potent inhibitor of the 11β-HSD-1 enzyme with reduced Pregnane-X receptor (PXR) transactivation activity. The binding orientation of this TZP series was revealed by X-ray crystallography structure studies. KEYWORDS: Enzyme inhibitors, human 11β-hydroxysteroid dehydrogenase-type 1, triazolopyridines

G

reported as a potential treatment for diabetes, metabolic syndrome, and related disorders.12−21 Our laboratory has recently reported novel inhibitors of 11βHSD-1 based on a 1,2,4-triazolopyridine (TZP) framework, represented by compound 1 (Figure 1), which showed reasonable good potency but with liabilities, such as PXR activity (EC50 = 1.2 μM). Structure−activity relationship (SAR) optimization of substituents at the C-3 and C-8 positions of the TZP core resulted in the identification of compound 2a as a reasonable potent and metabolically stable inhibitor of the

lucocorticoids such as cortisol are stress hormones that play an important role in regulating carbohydrate, protein, and lipid metabolism, as well as modulating inflammatory and immune responses. Glucocorticoids are present in two forms in humans: the active cortisol (corticosterone in rodents) and the inactive cortisone (11-dehydrocorticosterone in rodents). Excess cortisol expression in humans, as typified in patients with Cushing’s syndrome, can cause a variety of abnormalities including obesity, insulin resistance, hyperglycemia, dyslipidemia, and hypertension. These symptoms are also typically found in patients with metabolic syndrome or related diabetes/obesity diseases.1−3 A good correlation of salivary cortisol levels with components of the metabolic syndrome have been reported.4 Largely expressed in liver and adipose tissue, 11β-HSD-1 is an enzyme that catalyzes the conversion of inactive cortisone to the active glucocorticoid hormone cortisol.5 Transgenic mice that overexpress 11β-HSD-1 in the adipose develop many of the features of metabolic syndrome, including glucose intolerance, insulin resistance, dyslipidemia, hypertension, and obesity.6,7 In contrast, whole-body genetic knockout of 11β-HSD-1 expression in mice placed on a high fat diet has been shown to improve glucose tolerance and reduce triglyceride level relative to wild-type controls on a similar diet.8 These preclinical results suggest that inhibition of 11β-HSD-1 activity in humans may provide a beneficial impact for the treatment of metabolic syndrome. As a result, significant attention has been given to modulation of this enzyme from both academia and industry,9−11 and numerous selective inhibitors have been © XXXX American Chemical Society

Figure 1. Generation of lead 1,2,4-trizolopyridine (TZP) 11β-HSD-1 inhibitor 3b based on previous leads 1 and 2a. Received: April 9, 2014 Accepted: May 22, 2014

A

dx.doi.org/10.1021/ml500144h | ACS Med. Chem. Lett. XXXX, XXX, XXX−XXX

ACS Medicinal Chemistry Letters

Letter

enzyme without Pregnane-X receptor (PXR) activity.22 While certain aliphatic bicyclic rings (such as the bicyclo[2.2.2]octan1-ol in compound 2a) served well to address potency and metabolic stability issues for this chemotype, the rigidity and limited synthetic accessibility of these bridged structures made further modification to this moiety very challenging. Looking to incorporate simpler replacements for the bicyclic ring system, we turned our attention to incorporation of a 3-(1-phenyl)cyclopropyl functionality, which was previously identified as a potent binding pharmacophore in a related series of substituted triazoles.15,23 Indeed, replacement of the bicyclo[2.2.2]octan-1ol moiety in 2a with 4-chlorophenyl-cyclopropyl group resulted in 3b, a compound that possessed excellent inhibitory activity toward human 11β-HSD-1 (IC50 = 1.0 nM) while retaining good metabolic stability. Unfortunately the modification also resulted in a significantly increased liability for PXR transactivation, an off-target liability that was also observed in a series of related triazoles from Merck.15,16 As such we initiated a systematic SAR study to determine if a robust separation of PXR transactivation from dehydrogenase activity was possible in this series. Our initial SAR approach focused on modification of the lefthand portion of the molecule. A convenient synthesis of these derivatives is given in Scheme 1. The commercially available 1-

Table 1. SAR of 3-[1-(4-Chlorophenyl)cyclopropyl]-8aryloxy-TZP Analogues 3a−3la

compd

aryl

IC50a (nM)

PXR EC50 (μM)/Ymax (%)

3a 3b 3c 3d 3e 3f 3g 3h 3i 3j 3k 3l

Ph 2-Cl-Ph 3-Cl-Ph 4-Cl-Ph 2-F-Ph 2-(N-morpholinyl)-Ph 2-CF3O-Ph 2-hydroxyl-Ph 2-methylsulfonyl-Ph 2-Ph-Ph 2-naphthyl 2-Me-pyridin-3-yl

1.7 1.0 22 21 0.7 0.9 0.5 1.4 8.1 2.7 20 5.5

1.8/71 0.97/100 0.8/95 1.2/85 1.1/86 0.4/123 0.3/123 2.6/50 0.7/105 0.5/95 0.8/75 2.8/63

a IC50 values refer to biochemical human 11β-HSD-1 assay data; average of at least two replicates.

similar human 11β-HSD-1 inhibition potencies. The more polar hydroxyl group (3h) was also well tolerated, but the methylsulfonyl group (3i) exhibited attenuated in vitro activity. A phenyl (3j) at the ortho-position exhibited good potency, but fused 2-naphthalene analogue (3k) was much less active. While the majority of these analogues showed high human 11β-HSD1 inhibitory activity, all possessed significant PXR activity (EC50 < 3 μM). The more lipophilic analogues (e.g., 3g and 3f) were among the most potent tranasactivators in this series. Incorporation of polar groups (e.g., 3h and 3l) only had a modest effect on attenuating PXR transactivation. We next directed our attention to the incorporation of smaller aliphatic rings at the C-8 position of the TZP core. Introduction of cycloalkyl substituents was achieved via reaction of bromo-TZP intermediate 7 with various alkyl boronic acids or alkyl zinc halides under Pd-catalysis (Scheme 2). This method was used to generate compounds 9e−g in 40− 75% yields. Compound 9h was made directly from 2hydrazinyl-3-methylpyridine as shown in Scheme 1.

Scheme 1a

a

Reagents and conditions: (a) N-methylmorpholine, i-BuOCOCl, 0 °C, (90%); (b) PPh3Cl2, N,N-diisopropylethylamine, CH2Cl2, rt, (75%); (c) Cs2CO3, DMF or neat, 120 °C.

(4-chlorophenyl) cyclopropane-1-carboxylic acid (4) was coupled with hydrazine 5 to afford acyl hydrazide 6 in good yield (90%). Mild cyclodehydration of 6 using PPh3Cl2 produced bromo-triazolopyridine 7 in 75% yield. Coupling of 7 with various phenols 8 under modified literature conditions (i.e., Cs2CO3 under concentrated or neat reaction conditions) typically resulted in acceptable yields (>50%) of final products. Analogues 3a−l were synthesized via this route as well as analogues 9a−d with lower yield (5−20%). As outlined in Table 1, the presence of the ortho-Cl substituent 3b only marginally enhanced potency as compared to the unsubstituted phenyl analogue 3a. A chlorine was employed to study the positional effect of phenyl substitution. Compared with ortho-substituted phenyl analogue 3b, chlorine at the meta- and para-positions of the phenyl ring (3c and 3d, respectively) resulted in a significant loss of potency. This finding suggested that the ortho-position may be a favorable site to explore additional substituent effects on the phenyl ring. For this purpose, several functional groups with varied steric and electronic properties were introduced. Groups such as fluoro (3e), an electron-donating N-morpholinyl (3f), and the large hydrophobic trifluoromethoxy (3g, ClogP, 6.07) afforded

Scheme 2. Synthesis of Alkyl-TZP Analoguesa

a

Reagents and conditions: alkyl-B(OH)2 or alkyl-Zn-Br, 1,1′-bis(diphenylphosphino)ferrocene palladium(II)dichloride dichloromethane complex, K3PO4 or K2CO3, 85 °C, DMF.

Among the first analogues examined (Table 2) was the cyclohexyloxy derivative 9a, which was the aliphatic counterpart of compound 3a. However, compound 9a showed much weaker 11β-HSD-1 inhibition while retaining similar PXR activity. Attenuated potency was also observed for the cyclopropylmethoxy analogue 9c. Although the 4-tetrahydropyranyloxy substituent 9b (ClogP, 3.49) exhibited reduced 11βB

dx.doi.org/10.1021/ml500144h | ACS Med. Chem. Lett. XXXX, XXX, XXX−XXX

ACS Medicinal Chemistry Letters

Letter

triazole core and is the most closely resembled TZP core of interest (PDB code 3D5Q).24,25 While complete flexibility of the ligands was assumed in the calculation, the receptor was treated rigidly during docking, and the docking site was assumed to be the pocket occupied by the ligand in the 11βHSD-1 complex. As a post processing step, local energy minimization in the gas phase using the OPLS 2005 force field was carried out in order to relax the overall structure; this also allowed for partial flexibility of the receptor atoms surrounding the docked ligand. The favored poses from the docking calculations are shown in Figure 3 along with the residues in

Table 2. SAR of 3-[1-(4-Chlorophenyl)cyclopropyl]-TZP 9a−h with (Cyclo)alkyloxy and Cycloalkyloxy or Alkyl Groups at C-8a

compd

R

IC50a (nM)

PXR EC50 (μM)/Ymax (%)

9a 9b 9c 9d 9e 9f 9g 9h

cyclohexyloxy 4-tetrahydropyranyloxy c-PrCH2O cyclobutoxy cyclobutyl cyclopropyl isopropyl methyl

42 65 51 4.2 2.4 1.6 0.61 17

0.6/103 10/52 not tested 1.5/60 0.9/24 50/14 1.0/34 8.8/22

a

IC50 values refer to biochemical human 11β-HSD-1 assay data; average of at least two replicates.

HSD-1 inhibitory potency, the dramatically reduced PXR activation profile (EC50 = 10 μM, Ymax = 52%) again suggested that reducing lipophilicity at this position of the molecule was desired. A significant advance was observed by removing “O” linker affording compounds 9e−9h, which exhibited good to excellent 11β-HSD-1 potency while attenuating the full agonist profile (Ymax < 35%) for PXR. The “sweet-spot” in this SAR study is observed with compound 9f, which exhibits a superior combination of high enzyme inhibitory activity and low PXR transactivation (EC50 > 50 μM). Certain structure−activity and structure−liability relationships observed in this new TZP series (as exemplified by compounds 9b and 9f) appeared highly reminiscent with that observed in the lead TZP chemotype (as exemplified by compound 2a). The original assumption that the 1-(4chlorophenyl)cyclopropyl group in 3b was a surrogate for the [2,2,2]-bicyclooctanol group in 2a may now be better explained by an ∼180° rotation of the TZP core so that the 1-(4chlorophenyl)cyclopropyl group in 3b (now spatially represented by 3b′) is actually substituting for the 3-chloro-2methylphenoxy group in 2 instead (see Figure 2).

Figure 3. Preferred binding poses of compounds 2a and 3b using protein coordinates from 3D5Q, which has a 1,2,4-triazole ligand bound at the active site. The protein carbon atoms are shown in green and those of compound 2a and 3b in cyan; nitrogen atoms are shown in blue, oxygen atoms in red, and chlorine atoms in dark green. The interatomic distances (in Angstroms) between polar ligand atoms and protein residues that are indicative of hydrogen bonds are shown in magenta.

the binding pocket that contribute most significantly to the binding energy. The most striking observation is that the two compounds 2a and 3b bind to 11β-HSD-1 in opposite orientations, as inferred from our SAR studies. As with the triazole structure 3D5Q, both TZPs bind in an orientation in which the triazole ring is oriented toward the side chains of Ser170 and Tyr183 suggesting probable hydrogen bonds. The overall docking pose shows that the phenoxy substituent at the 8 position in the TZP scaffold lies on opposite sides of the binding pocket in compounds 2a and 3b. As seen in Figure 3A, the preferred tendency of 2a to bind in an orientation opposite to 3b may be caused by the tendency of the hydroxyl group on the bicyclo[2.2.2]octyl ring in the former to form a hydrogen bond with the backbone carbonyl oxygen on Thr124. The interaction energy of this hydrogen bond is estimated at about 1.5 kcal/mol from the docking result, favoring this pose over the 180-degree flipped pose (as for compound 3b). The opposite binding orientations based on docking suggests that while analogues of compound 2a (lead TZP series) will bind in the orientation shown for 2a, analogues of compound 3b (alternate TZP series), which besides the two hydrogen bond interactions shown in Figure 3B does not show any other polar interactions between ligand and protein, may often bind in an orientation similar to 2a rather than 3b depending on the nature of the substitutions in the two terminal phenyl rings. This is consistent with the observation that the SAR for C-3 and C-8 substitutions on the TZP core do not parallel for the lead and alternate series. The observed SAR findings and docking calculation predictions prompted us to further examine the binding mode of these compounds to 11β-HSD-1 using X-ray crystallography to confirm the binding orientation. Although X-ray crystal structures are not available for a human 11β-HSD-

Figure 2. Reversal of core binding orientations for “original” TZP series 2a versus “alternate” TZP series 9b.

In order to visualize binding modes for compounds in the two TZP subseries, compounds 2a and 3b were docked into a crystal structure of 11β-HSD-1 complexed with NADPH and a ligand, 3-[1-(4-fluorophenyl)cyclopropyl]-4-(1-methylethyl)5-[4-(trifluoromethoxy)phenyl]-4H-1,2,4-triazole (structure shown in Supporting Information), which contains a 1,2,4 C

dx.doi.org/10.1021/ml500144h | ACS Med. Chem. Lett. XXXX, XXX, XXX−XXX

ACS Medicinal Chemistry Letters

Letter

superposition of compounds 2b and 9f and surrounding residues are shown in Figure 5D. These findings are consistent with previously reported ∼180° flips of inhibitors of 11β-HSD1 derived from different chemical series by others,28 and further revealed the flexibility of 11β-HSD-1 inhibitor of binding mode. As compound 9f seemed to afford a superior combination of potency, low PXR activity, and reasonable solubility (76 μg/ mL, pH 7.4), it was selected for more extensive in vitro and in vivo evaluation. As observed with other analogues in this series, compound 9f was essentially inactive versus 11β-HSD-2 (IC50 > 10 μM), an unwanted activity. With the exception of CYP2C19 (IC50 ≈ 2 μM), compound 9f was not a significant inhibitor of other human CYP450 enzymes such as 1A2, 2C8, 2C9, 2D6, or 3A4 (IC50 > 20 μM). Ion channel activities including hERG were acceptable (hERG flux IC50 > 60 μM). Excellent in vitro metabolic stability in isolated microsomes was observed across all species tested (human, mouse, cyno, and rat: ∼100% remaining after 30 min of incubation). When orally dosed to male Balb/C mice at 3.5 mpk (0.5% methocel/0.1% Tween-80 in water as the vehicle), oral absorption was rapid with a short tmax (1 h). The maximum plasma concentration cmax was 2.5 μM, and plasma exposure was acceptable with the AUC (0−8 h) reaching 5.6 μM·h. The compound was found to distribute extensively in adipose tissue as well, affording an adipose/plasma ratio of ∼5 when measured 8 h postdosing. Although compound 9f demonstrated excellent human 11βHSD-1 inhibition (IC50 = 1.6 nM), its activity was dramatically weaker for the murine enzyme (IC50 = 241 nM). A significant difference in potency with respect to species was also observed comparing activities in human (HEK IC50 = 13 nM) and mouse (3T3L1 IC50 = 293 nM) derived cellular assays. Indeed, attenuated activity for mouse and rat (versus human or cyno) 11β-HSD-1 was typically consistent across the chemotype and complicated in vivo testing of analogues with this series. As such, it was not surprising that compound 9f failed to elicit a response in a standard DHC challenge assay in mice at doses up to 30 mpk. In summary, a series of novel, potent, and selective inhibitors of human 11β-HSD-1 enzyme were revealed through structure−activity relationship studies of a 1,2,4-triazolopyridine (TZP) core, and representative lead compounds such as 9f were identified. A disconnect in the SAR between analogues in this series (represented by compounds like 3e and 9f) and the lead TZP analogue 2a suggested a “flipped” binding orientation within the catalytic site of 11β-HSD-1, which was confirmed by docking calculations and X-ray structure studies. This finding suggests that the TZP core might provide an ideal, flexible platform for future compound optimization. While these compounds proved to be challenging to evaluate due to their weak rodent activities, they served as excellent starting points for further modifications of this chemotype resulting from discovery of our clinical candidate, which will be reported in due course.

1 complex with either compound 2a or 3b, the complex structures of closely related analogues 2b and 3e (Figure 4)

Figure 4. Structures and human 11β-HSD-1 activities of compounds 2b and 3e.

Figure 5. Binding of compounds to human 11β-HSD-1 as obtained through X-ray crystallographic analysis. Figures 5A (2b), B (3e), and C (9f) show the final coordinates with the initial (prior to placement) 2Fo − Fc electron density (magenta; contoured at 1 rmsd) and several surrounding residues. Carbon atoms of compounds are shown in green and those of 11β-HSD-1 in cyan. Nitrogen atoms are shown in blue, oxygen atoms in red, fluorine atoms in teal, and chlorine atoms in pea green. Hydrogen bonds are shown as a series of ellipsoids (black). In particular, note that the two exposed nitrogen atoms of the triazolyl moiety form hydrogen bonds to Ser 170 OH and Tyr 183 OH. Figure 5D depicts the superposition of compounds 2b (cyan) and 9f (magenta) and surrounding residues showing that the orientation of the core triazolopyridine flips ∼180° about the vertical axis while maintaining the hydrogen bonds to Ser 170 OH and Tyr 183 OH. Note also that the o-fluorophenyl moiety of compound 2b forces Tyr 177 side chain to flip out of the way.

were obtained at 2.35 Å resolution (Figure 5).26,27 The X-ray crystal structures confirmed our hypothesis: the orientation of the core TZP rings are indeed rotated ∼180° about the vertical axis for the two compounds as shown in Figure 2 while maintaining productive hydrogen bonding interactions to the Ser 170 and Tyr 183 hydroxyl groups (compare Figure 5A,B). Furthermore, compound 9f was also shown to exhibit the same orientation as that of 3e (Figure 5C). It is worthy to point out that the phenylcyclopropyl group in the 3D5Q ligand is oriented toward T124, whereas in 3e and 9f it is oriented in the opposite direction, toward Y177, as determined by the preferred binding orientation. For direct comparison, the



ASSOCIATED CONTENT

* Supporting Information S

Protocols or methods of in vitro and in vivo assays, analytical and spectroscopic data for compounds 3−9h, and X-ray crystallographic data of compound 2b, 3e, and 9f. This material is available free of charge via the Internet at http://pubs.acs.org. D

dx.doi.org/10.1021/ml500144h | ACS Med. Chem. Lett. XXXX, XXX, XXX−XXX

ACS Medicinal Chemistry Letters



Letter

dehydrogenase type 1: Discovery of PF-915275. Bioorg. Med. Chem. Lett. 2009, 19, 3493−3497. (14) Rosenstock, J.; Banarer, S.; Fonseca, V. A.; Inzucchi, S. E.; Sun, W.; Yao, W.; Hollis, G.; Flores, R.; Levy, R.; Williams, W.; Seckl, J. R.; Huber, R. The 11β-hydroxysteroid dehydrogenase type 1 inhibitor INCB13739 improves hyperglycemia in patients with type 2 diabetes inadequately controlled by metformin monotherapy. Diabetes Care 2010, 33, 1516−1522. (15) Shah, S.; Hermanowski-Vosatka, A.; Gibson, K.; Ruck, R. A.; Jia, G.; Zhang, J.; Hwang, P. M. T.; Ryan, N. W.; Langdon, R. B.; Feig, P. U. Efficacy and safety of the selective 11β-HSD-1 inhibitors MK-0736 and MK-0916 in overweight and obese patients with hypertension. J. Am. Soc. Hypertens. 2011, 5, 166−176. (16) Sun, W.; Maletic, M.; Mundt, S. S.; Shah, K.; Zokian, H.; Lyons, K.; Waddell, S. T.; Balkovec, J. Substituted phenyl triazoles as selective inhibitors of 11β-hydroxysteroid dehydrogenase type 1. Bioorg. Med. Chem. Lett. 2011, 21, 2141−2145. (17) Xu, Z.; Tice, C. M.; Zhao, W.; Cacatian, S.; Ye, Y.; Singh, S. B.; Lindblom, P.; McKeever, B. M.; Krosky, P. M.; Kruk, B. A.; Berbaum, J.; Harrison, R. K.; Johnson, J. A.; Bukhtiyarov, Y.; Panemangalore, R.; Scott, B. B.; Zhao, Y.; Bruno, J. G.; Togias, J.; Guo, J.; Guo, R.; Carroll, P. J.; McGeehan, G. M.; Zhuang, L.; He, W.; Claremon, D. A. Structure-based design and synthesis of 1,3-oxazinan-2-one inhibitors of 11β-hydroxysteroid dehydrogenase type 1. J. Med. Chem. 2011, 54, 6050−6062. (18) Scott, J. S.; Bowker, S. S.; deSchoolmeester, J.; Gerhardt, S.; Hargreaves, D.; Kilgour, E.; Lloyd, A.; Mayers, R. M.; McCoull, W.; Newcombe, N. J.; Ogg, D.; Packer, M. J.; Rees, A.; Revill, J.; Schofield, P.; Selmi, N.; Swales, J. G.; Whittamore, P. R. O. Novel acidic 11βhydroxysteroid dehydrogenase type 1 (11β-HSD1) inhibitor with reduced acyl glucuronide liability: The discovery of 4-[4-(2adamantylcarbamoyl)-5-tert-butyl-pyrazol-1-yl]benzoic acid (AZD8329). J. Med. Chem. 2012, 55, 10136−10147. (19) Yu, J.; Liu, H.; Xia, G.; Liu, L.; Xu, Z.; Chen, Q.; Ma, C.; Sun, X.; Xu, J.; Li, H.; Li, P.; Shi, Y.; Xiong, B.; Liu, X.; Shen, J. Discovery of 2-Alkyl-1-arylsulfonylprolinamides as 11β-hydroxysteroid dehydrogenase type 1 Inhibitors. ACS Med. Chem. Lett. 2012, 3, 793−798. (20) Wan, Z.-K.; Chenail, E.; Li, H.; Ipek, M.; Xiang, J.; Suri, V.; Hahm, S.; Bard, J.; Svenson, K.; Xu, X.; Tian, X.; Wang, M.; Li, X.; Johnson, C. E.; Qadri, A.; Panza, D.; Perreault, M.; Mansour, T. S.; Tobin, J. F.; Saiah, E. Discovery of HSD-621 as a potential agent for the treatment of type 2 diabetes. ACS Med. Chem. Lett. 2013, 2, 118− 123. (21) Bauman, D. R.; Whitehead, A.; Contino, L. C.; Cui, J.; GarciaCalvo, M.; Gu, X.; Kevin, N.; Ma, X.; Pai, L.; Shah, K.; Shen, X.; Stribling, S.; Zokian, H. J.; Metzger, J.; Shevell, D. E.; Waddell, S. T. Evaluation of selective inhibitors of 11β-HSD1 for the treatment of hypertension. Bioorg. Med. Chem. Lett. 2013, 23, 3650−3653. (22) Wang, H.; Robl, J. A.; Hamann, L. G.; Simpkins, L. M.; Golla, R.; Li, X.; Seethala, R.; Zvyaga, T.; Gordon, D. A.; Robl, J. A.; Li, J. L. Generation of 3,8-substituted 1,2,4-triazolopyridines as potent inhibitors of human 11β-hydroxysteroid dehydrogenase type-1. Bioorg. Med. Chem. Lett. 2011, 21, 4146−4149. (23) Zhu, Y.; Olson, S. H.; Graham, D.; Patel, G.; HermanowskiVosatka, A.; Mundt, S.; Shah, K.; Springer, M. Phenylcyclobutyl triazoles as selective inhibitors of 11β-hydroxysteroid dehydrogenase type I. Bioorg. Med. Chem. Lett. 2008, 18, 3412−3416. (24) The software used for this purpose was Glide SP as implemented in Maestro verson 9.0 (Glide, version 5.7, Schrödinger, LLC, New York, NY, 2011. Friesner, R. A.; Banks, J. L.; Murphy, R. B.; Halgren, T. A.; Klicic, J. J.; Mainz, D. T.; Repasky, M. P.; Knoll, E. H.; Shaw, D. E.; Shelley, M.; Perry, J. K.; Francis, P.; Shenkin, P. S. Glide: A new approach for rapid, accurate docking and scoring. 1. Method and assessment of docking accuracy. J. Med. Chem. 2004, 47, 1739− 1749. (25) Tu, H.; Powers, J. P.; Liu, J.; Ursu, S.; Sudom, A.; Yan, X.; Xu, H.; Meininger, D.; Degraffenreid, M.; He, X.; Jaen, J. C.; Sun, D.; Labelle, M.; Yamamoto, H.; Shan, B.; Walker, N. P.; Wang, Z. Distinctive molecular inhibition mechanisms for selective inhibitors of

AUTHOR INFORMATION

Corresponding Authors

*(J.L.) Tel: 609-818-7123. E-mail: [email protected]. *(J.A.R.) Tel: 609-818-5048. E-mail: jeff[email protected]. Notes

The authors declare no competing financial interest.

■ ■

ABBREVIATIONS USED 11β-HSD-1, 11β-hydroxysteroid dehydrogenase type 1; TZP, triazolopyridines; PXR, Pregnane-X receptor REFERENCES

(1) Tomlinson, J. W.; Stewart, P. M. Mechanisms of disease: selective inhibition of 11β-hydroxysteroid dehydrogenase type 1 as a novel treatment for the metabolic syndrome. Nat. Clin. Practice Endocrinol. Metab. 2005, 1, 92−99. (2) Gathercole, L. L.; Lavery, G. G.; Morgan, S. A.; Cooper, M. S.; Sinclair, A. J.; Tomlinson, J. W.; Stewart, P. M. 11β-Hydroxysteroid dehydrogenase 1: translational and therapeutic aspects. Endocrine Rev. 2013, 34 (4), 525−555. (3) Morton, N. M. Obesity and corticosteroids: 11β-hydroxysteroid type 1 as a cause and therapeutic target in metabolic disease. Mol. Cell. Endocrinol. 2010, 316, 154−164. (4) Oltmanns, K. M.; Dodt, B.; Schultes, B.; Raspe, H. H.; Schweiger, U.; Born, J.; Fehm, H. L.; Peters, A. Cortisol correlates with metabolic disturbances in a population study of type 2 diabetic patients. Eur. J. Endocrinol. 2006, 154, 325−331. (5) Jamieson, P. M.; Walker, B. R.; Chapman, K. E.; Rossiter, S.; Seckl, J. R. 11β-Hydroxysteroid dehydrogenase type 1 is a predominant 11β reductase in the intact perfused rat liver. J. Endocrinol. 2000, 165, 685−692. (6) Masuzaki, H.; Paterson, J.; Shinyama, H.; Morton, N. M.; Mullins, J. J.; Seckl, J. R.; Flier, J. S. A transgenic model of visceral obesity and the metabolic syndrome. Science 2001, 294, 2166−2170. (7) Masuzaki, H.; Yamamoto, H.; Kenyon, C. J.; Elmquist, J. K.; Morton, N. M.; Paterson, J. M.; Shinyama, H.; Sharp, M. G. F.; Fleming, S.; Mullins, J. J.; Seckl, J. R.; Flier, J. S. Transgenic amplification of glucocorticoid action in adipose tissue causes high blood pressure in mice. J. Clin. Invest. 2003, 112, 83−90. (8) Morton, N. M.; Holmes, M. C.; Fievet, C.; Staels, B.; Tailleux, A.; Mullins, J. J.; Seckl, J. R. Improved lipid and lipoprotein profile, hepatic insulin sensitivity, and glucose tolerance in 11β-hydroxysteroid dehydrogenase type 1 null mice. J. Biol. Chem. 2001, 276, 41293− 41300. (9) Fotsch, C.; Wang, M. Blockade of glucocorticoid excess at the tissue level: inhibitors of 11β-hydroxysteroid dehydrogenase type 1 as a therapy for type 2 diabetes. J. Med. Chem. 2008, 51, 4851−4857. (10) Boyle, C. D.; Kowalski, T. J. 11β-Hydroxysteroid dehydrogenase type 1 inhibitors: a review of recent patents. Expert Opin. Ther. Pat. 2009, 19, 801−825. (11) Ge, R.; Huang, Y.; Liang, G.; Li, X. 11β-Hydroxysteroid dehydrogenase type 1 inhibitors as promising therapeutic drugs for diabetes: status and development. Curr. Med. Chem. 2010, 17, 412− 422. (12) Veniant, M. M.; Hale, C.; Hungate, R. W.; Gahm, K.; Emery, M. G.; Jona, J.; Joseph, S.; Adams, J.; Hague, A.; Moniz, G.; Zhang, J.; Bartberger, M. D.; Li, V.; Syed, R.; Jordan, S.; Komorowski, R.; Chen, M. M.; Cupples, R.; Kim, K. W.; St. Jean, D. J., Jr.; Johansson, L.; Henriksson, M. A.; Williams, M.; Vallgarda, J.; Fotsch, C.; Wang, M. Discovery of a potent, orally active 11β-hydroxysteroid dehydrogenase type 1 inhibitor for clinical study: Identification of (S)-2-((1S,2S,4R)bicyclo[2.2.1]heptan-2-ylamino)-5-isopropyl-5-methylthiazol-4(5H)one (AMG 221). J. Med. Chem. 2010, 53, 4481−4187. (13) Siu, M.; Johnson, T. O.; Wang, Y.; Nair, S. K.; Taylor, W. D.; Cripps, S. J.; Matthews, J. J.; Edwards, M. P.; Pauly, T. A.; Ermolieff, J.; Castro, A.; Hosea, N. A.; LaPaglia, A.; Fanjul, A. N.; Vogel, J. E. N(Pyridin-2-yl) arylsulfonamide inhibitors of 11β-hydroxysteroid E

dx.doi.org/10.1021/ml500144h | ACS Med. Chem. Lett. XXXX, XXX, XXX−XXX

ACS Medicinal Chemistry Letters

Letter

human 11β-hydroxysteroid dehydrogenase type 1. Bioorg. Med. Chem. Lett. 2008, 16, 8922−8931. (26) Emsley, P.; Lokhamp, B.; Scott, W.; Cowtan, K. Features and development of coot. Acta Crystallogr., Sect. D: Biol. Crystallogr. 2010, 66, 486−501. (27) PDB codes of 11β-HSD1 complexes are the following: 2b (4IJU), 3e (4IJV), and 9f (4IJW). (28) Johansson, L.; Fotsch, C.; Bartberger, M. D.; Castro, V. M.; Chen, M.; Emery, M.; Gustafsson, S.; Hale, C.; Hickman, D.; Homan, E.; Jordan, S. R.; Komorowski, R.; Li, A.; McRae, K.; Moniz, G.; Matsumoto, G.; Orihuela, C.; Palm, G.; Veniant, M.; Wang, M.; Williams, M.; Zhang, J. 2-Amino-1,3-thiazol-4(5H)-ones as potent and selective 11β-hydroxysteroid dehydrogenase type 1 inhibitors: Enzyme-ligand co-crystal structure and demonstration of pharmacodynamic effects in C57Bl/6 mice. J. Med. Chem. 2008, 51, 2933−2943.

F

dx.doi.org/10.1021/ml500144h | ACS Med. Chem. Lett. XXXX, XXX, XXX−XXX